EP2856161B1 - Essais - Google Patents

Essais Download PDF

Info

Publication number
EP2856161B1
EP2856161B1 EP13734141.8A EP13734141A EP2856161B1 EP 2856161 B1 EP2856161 B1 EP 2856161B1 EP 13734141 A EP13734141 A EP 13734141A EP 2856161 B1 EP2856161 B1 EP 2856161B1
Authority
EP
European Patent Office
Prior art keywords
membrane protein
label
donor
acceptor
protein
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
EP13734141.8A
Other languages
German (de)
English (en)
Other versions
EP2856161A2 (fr
Inventor
Fiona Hamilton Marshall
Seyed Ali Jazayeri-Dezfuly
Jayesh Chhotubhai PATEL
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Nxera Pharma UK Ltd
Original Assignee
Heptares Therapeutics Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Heptares Therapeutics Ltd filed Critical Heptares Therapeutics Ltd
Publication of EP2856161A2 publication Critical patent/EP2856161A2/fr
Application granted granted Critical
Publication of EP2856161B1 publication Critical patent/EP2856161B1/fr
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6803General methods of protein analysis not limited to specific proteins or families of proteins
    • G01N33/6842Proteomic analysis of subsets of protein mixtures with reduced complexity, e.g. membrane proteins, phosphoproteins, organelle proteins
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/536Immunoassay; Biospecific binding assay; Materials therefor with immune complex formed in liquid phase
    • G01N33/542Immunoassay; Biospecific binding assay; Materials therefor with immune complex formed in liquid phase with steric inhibition or signal modification, e.g. fluorescent quenching
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/72Receptors; Cell surface antigens; Cell surface determinants for hormones
    • C07K14/723G protein coupled receptor, e.g. TSHR-thyrotropin-receptor, LH/hCG receptor, FSH receptor
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N21/00Investigating or analysing materials by the use of optical means, i.e. using sub-millimetre waves, infrared, visible or ultraviolet light
    • G01N21/62Systems in which the material investigated is excited whereby it emits light or causes a change in wavelength of the incident light
    • G01N21/63Systems in which the material investigated is excited whereby it emits light or causes a change in wavelength of the incident light optically excited
    • G01N21/64Fluorescence; Phosphorescence
    • G01N21/6428Measuring fluorescence of fluorescent products of reactions or of fluorochrome labelled reactive substances, e.g. measuring quenching effects, using measuring "optrodes"
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/566Immunoassay; Biospecific binding assay; Materials therefor using specific carrier or receptor proteins as ligand binding reagents where possible specific carrier or receptor proteins are classified with their target compounds
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/705Assays involving receptors, cell surface antigens or cell surface determinants
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2458/00Labels used in chemical analysis of biological material

Definitions

  • the present invention relates to assays for assessing the stability of a membrane protein, especially a G protein-coupled receptor (GPCR).
  • GPCR G protein-coupled receptor
  • membrane proteins should be stable for many days in any given detergent solution, but the detergents that are best suited to growing diffraction-quality crystals tend to be the most de-stabilising detergents i.e. those with short aliphatic chains and small or charged head groups. It is also the structures of human membrane proteins that we would like to solve, because these are required to help the development of therapeutic agents by the pharmaceutical industry. There is thus an overwhelming need to develop a generic strategy that will allow the production of detergent-stable eukaryotic integral membrane proteins for crystallisation and structure determination and potentially for other purposes such as drug screening, bioassay and biosensor applications.
  • GPCRs constitute a very large family of proteins that control many physiological processes and are the targets of many effective drugs. Accordingly, they are of considerable pharmacological importance. A list of GPCRs is given in Foord et al (2005) Pharmacol Rev. 57, 279-288 . GPCRs are generally unstable when isolated, and despite considerable efforts, few crystal structures exist.
  • GPCRs are druggable targets, and reference is made particularly to Overington et al (2006) Nature Rev. Drug Discovery 5, 993-996 which indicates that over a quarter of present drugs have a GPCR as a target.
  • GPCRs are thought to exist in multiple distinct conformations which are associated with different pharmacological classes of ligand such as agonists and antagonists, and to cycle between these conformations in order to function ( Kenakin T. (1997) Ann N Y Acad Sci 812, 116-125 ).
  • GPCRs have evolved to cycle through at least two distinct conformations, the agonist-bound form and the antagonist-bound form, and changes between these two conformations can occur spontaneously in the absence of ligand. It is thus likely that any purified receptors populate a mixture of conformations. Just adding ligands to GPCRs during crystallisation trials has not resulted in their structure determination.
  • StaRs stabilised receptors
  • the method of StaR generation results in the stabilisation of a specific conformation of receptor.
  • a detailed description of the method can be found elsewhere [2, 3, 4] and in WO 2008/114020 and WO 2009/071914 .
  • the solubilised receptor is incubated with an excess amount of either the agonist or antagonist, depending on the desired conformation.
  • the receptor/ligand complex is then heated at various temperatures for a certain length of time. Following the heating, the excess unbound ligand is separated from receptor/ligand complexes.
  • the percentage of ligand binding at any given temperature is used as readout for the amount of folded receptor remaining following heating. Plotting this data against temperature produces a thermal decay curve and the Tm value is defined as the temperature at which 50% of receptor activity is retained. This process relies on the availability of a good ligand with high affinity and favourable properties in detergent. However, in some cases such a ligand is not available and so an assay that circumvents the need for a ligand, i.e. where ligand binding activity is not used to measure receptor activity, is highly desirable.
  • Knepp et al (Biochemistry 50(4): 502-11, 2011 ) describe the use of Fluorescence Resonance Energy Transfer (FRET) [6] to assess membrane protein stability.
  • FRET Fluorescence Resonance Energy Transfer
  • donor and acceptor fluorophores must have well separated emission spectra, but the emission spectrum of the donor must overlap with the excitation spectrum of the acceptor fluorophore.
  • binding of a monoclonal antibody labelled with a FRET acceptor is used as a marker of GPCR denaturation.
  • a FRET signal is generated when the FRET acceptor is in the proximity of the FRET donor attached to the C-terminus of the GPCR.
  • a significant limitation of this method is that it requires the availability of suitable antibodies specific for the membrane protein in question.
  • the method of Knepp et al is unlikely to identify stabilising mutations that happen to be in the antibody binding site since these mutations will reduce or abolish the antibody binding. Additionally, mutations in the antibody binding site may increase the affinity of the antibody for the receptor without necessarily increasing conformational stability, thereby leading to false positives.
  • Another disadvantage of Knepp et al 's method is that the antibody may have a specific effect on the pharmacology of the GPCR. For example, the antibody may be a particular conformation of the GPCR that is not desired.
  • Protein aggregation upon unfolding is a general feature of all proteins that occurs to varying degrees in different proteins. It is accepted that the overall structural stability of a protein is inversely proportional to the levels of aggregation. Membrane proteins are known to exhibit high levels of aggregation after solubilisation and more so after denaturation.
  • Pollitt et al Neuron 40: 685-694, 2003
  • US 7,803,559 describe a cell-based assay to measure intracellular polyglutamine protein aggregation using FRET, and discuss its use in screening for aggregation regulators that may have therapeutic application in neurodegenerative diseases.
  • Roberti et al (Nature Methods 4(4): 345, 2007 ) and Rajan et al (PNAS 98(23): 13060-13065 ) describe FRET based methods for assessing the aggregation of proteins that are known to form aggregates as part of a disease pathology.
  • EP 2 450 445 describes a method for selecting a G-protein coupled receptor (GPCR) with increased stability, the method comprising (a) providing one or more mutants of a parent GPCR, (b) selecting a ligand, the ligand being one which binds to the parent GPCR when the GPCR is residing in a particular conformation, (c) determining whether the or each mutant GPCR has increased stability with respect to binding the selected ligand compared to the stability of the parent GPCR with respect to binding that ligand, and (d) selecting those mutants that have an increased stability compared to the parent GPCR with respect to binding the selected ligand.
  • GPCR G-protein coupled receptor
  • the present inventors have now identified methods for directly assessing aggregation of membrane proteins. Such methods allow one to identify the conditions and mutations that can minimise overall aggregation of the proteins. Compared to methods that indirectly measure aggregation such as that of Knepp et al, the present methods are believed to identify more mutations and conditions that can minimise aggregation of proteins. This is because some mutations and conditions may reduce protein aggregation without changing the structure of the protein in a way that would lead to a change in ligand binding or antibody binding, as is relied on as a read out for aggregation in the indirect methods.
  • an assay for assessing the conformational stability of a membrane protein comprising:
  • aggregation of the membrane proteins from the first and second populations is detected by virtue of the donor labels and acceptor labels coming into proximity with each other, and thereby generating a detectable signal, when the membrane proteins aggregate. If the membrane proteins are stable, there is less aggregation upon exposure to a destabilising agent and/or condition and so less signal, whereas if the membrane proteins are unstable, there is more aggregation upon exposure to a destabilising agent and/or condition and so more signal.
  • conformational stability we include the meaning of the stability of a particular conformation of a membrane protein (eg GPCR).
  • the stability of a particular conformation refers to how well that particular conformation can retain its structure when exposed to a denaturant or denaturing conditions.
  • a membrane protein with high conformational stability will have an extended lifetime of a particular conformation compared to the lifetime of a particular conformation of a membrane protein with low conformational stability.
  • denaturants/denaturing conditions include heat, detergent, a chaotropic agent and an extreme of pH, as described further below. As is well known in the art, such denaturants or denaturing conditions can affect secondary and tertiary structures of a protein but not the primary sequence.
  • membrane protein we mean a protein that is attached to or associated with a membrane of a cell or organelle.
  • the membrane protein is an integral membrane protein that is permanently integrated into the membrane and can only be removed using detergents, non-polar solvents or denaturing agents that physically disrupt the lipid bilayer.
  • the membrane protein is a GPCR.
  • Suitable GPCRs for use in the practice of the invention include, but are not limited to chemokine receptor, (eg CCR5), ⁇ -adrenergic receptor, adenosine receptor (eg A 2a receptor), and neurotensin receptor (NTR).
  • chemokine receptor eg CCR5
  • ⁇ -adrenergic receptor eg A 2a receptor
  • NTR neurotensin receptor
  • Other suitable GPCRs are well known in the art and include those listed in Hopkins & Groom supra.
  • the International Union of Pharmacology produce a list of GPCRs ( Foord et al (2005) Pharmacol. Rev. 57, 279-288 , and this list is periodically updated at http://www.iuphar-db.org/GPCR/ReceptorFamiliesForward).
  • GPCRs are divided into different classes, principally based on their amino acid sequence similarities. They are also divided into families by reference to the natural ligands to which they bind. All GPCRs are
  • the GPCR may be any of an adenosine receptor, a ⁇ -adrenergic receptor, a neurotensin receptor, a muscarinic acid receptor, a 5-hydroxytryptamine receptor, an adrenoceptor, an anaphylatoxin receptor, an angiotensin receptor, an apelin receptor, a bombesin receptor, a bradykinin receptor, a cannabinoid receptor, a chemokine receptor, a cholecystokinin receptor, a dopamine receptor, an endothelin receptor a free fatty acid receptor, a bile acid receptor, a galanin receptor, a motilin receptor, a ghrelin receptor, a glycoprotein hormone receptor, a GnRH receptor, a histamine receptor, a KiSS1-derived peptide receptor, a leukotriene and lipoxin receptor, a lysophospholipid receptor, a melanin-concentrating hormone
  • the GPCR may also be selected from any of the GPCRs listed in Table A hereinafter.
  • membrane proteins for which the methods of the invention may usefully be employed to measure stability include the orphan receptors listed in Table B hereinafter.
  • the amino acid sequences (and the nucleotide sequences of the cDNAs which encode them) of many membrane proteins (eg GPCRs) are readily available, for example by reference to GenBank.
  • Foord et al supra gives the human gene symbols and human, mouse and rat gene IDs from Entrez Gene ( http://www.ncbi.nlm.nih.gov/entrez ).
  • the amino acid sequences of human membrane proteins (eg GPCRs) can be deduced therefrom.
  • the membrane protein (eg GPCR) may be derived from any source, it is particularly preferred if it is from a eukaryotic source.
  • the membrane protein eg GPCR
  • a cDNA or gene was originally obtained using genetic material from the source, but that the protein may be expressed in any host cell (eg prokaryotic or eukaryotic host cell) subsequently.
  • a eukaryotic membrane protein eg GPCR
  • a prokaryotic host cell such as E .
  • the membrane protein may be composed of more than one different subunit.
  • the calcitonin gene-related peptide receptor requires the binding of a single transmembrane helix protein (RAMP1) to acquire its physiological ligand binding characteristics.
  • RAMP1 transmembrane helix protein
  • effector, accessory, auxiliary or GPCR-interacting proteins which combine with the GPCR to form or modulate a functional complex are well known in the art and include, for example, receptor kinases, G-proteins and arrestins ( Bockaert et al (2004) Curr Opinion Drug Discov and Dev 7, 649-657 ).
  • the membrane proteins eg GPCRs
  • the membrane proteins may be prepared by any suitable method.
  • the membrane protein is encoded by a suitable nucleic acid molecule and expressed in a suitable host cell.
  • Suitable nucleic acid molecules encoding the membrane protein eg GPCR
  • GPCR GPCR
  • Suitable expression systems include constitutive or inducible expression systems in bacteria or yeasts, virus expression systems such as baculovirus, semliki forest virus and lentiviruses, or transient transfection in insect or mammalian cells.
  • Suitable host cells include E .
  • Suitable animal host cells include HEK 293, COS, S2, CHO, NSO, DT40 and so on. It is known that some membrane proteins (eg GPCRs) require specific lipids (eg cholesterol) to function. In that case, it is desirable to select a host cell which contains the lipid. Additionally or alternatively the lipid may be added during isolation and purification of the membrane protein. It may also be desirable to add a ligand of the membrane protein (eg GPCR) as explained further below.
  • GPCR membrane proteins
  • the sample comprising the first population and second population of a membrane protein may be any suitable sample that contains the two populations.
  • population we include a plurality of the same specific type of membrane protein, as opposed to a mixture of different proteins.
  • the population may comprise at least 2, 5, 10, 50, 100, 200, 500, 1000, 5000, 10000, 100000, 10 6 , 10 7 , 10 8 , 10 9 , 10 10 , 10 11 , 10 12 , 10 13 or 10 14 protein molecules, or more. It will be appreciated that the precise number of protein molecules may depend on the expression level of the particular membrane protein and this may differ from protein to protein.
  • the populations of membrane protein are provided in a suitable solubilised form in which the membrane proteins maintain structural integrity and are in a functional form (eg are able to bind ligand).
  • An appropriate solubilising system such as a suitable detergent (or other amphipathic agent) and buffer system is used, which may be chosen by the person skilled in the art to be effective for the particular protein.
  • Typical detergents which may be used include, for example, dodecylmaltoside (DDM) or CHAPS or octylglucoside (OG) or many others.
  • the sample comprises one or more detergents selected from DDM; C 11 -, C 10 -, C 9 - or C 8 - maltoside or glucoside; LDAO; and SDS. It may be convenient to include other compounds such as cholesterol hemisuccinate or cholesterol itself or heptane-1,2,3-triol. The presence of glycerol or proline or betaine may be useful. It is important that the membrane protein (eg GPCR), once solubilised from the membrane in which it resides, must be sufficiently stable to be assayed. For some membrane proteins (eg GPCRs), DDM will be sufficient, but glycerol or other polyols may be added to increase stability for assay purposes, if desired.
  • DDM will be sufficient, but glycerol or other polyols may be added to increase stability for assay purposes, if desired.
  • Further stability for assay purposes may be achieved, for example, by solubilising in a mixture of DDM, CHAPS and cholesterol hemisuccinate, optionally in the presence of glycerol.
  • solubilising in a mixture of DDM, CHAPS and cholesterol hemisuccinate optionally in the presence of glycerol.
  • glycerol eg GPCRs
  • Nanodiscs may also be used for solubilising extremely unstable membrane proteins in a functional form.
  • the membrane protein is provided in a crude extract (eg of the membrane fraction from the host cell in which it has been expressed, such as E . coli or HEK293T cells). It may be provided in a form in which the membrane protein comprises at least 75%, or at least 80% or 85% or 90% or 95% or 98% or 99% of the protein present in the sample. Alternatively, the membrane protein may be provided in a semi-purified form as described in Example 1 wherein the membrane protein of interest is not the most abundant species in the sample.
  • the membrane protein may be provided in a sample where only 5-50% of the total protein in the sample is the membrane protein (eg at least 5% or 10% or 15% or 20% or 25% or 30% or 35% or 40% or 45% of the protein present in the sample).
  • the membrane protein may be provided in a sample where only 5-50% of the total protein in the sample is the membrane protein (eg at least 5% or 10% or 15% or 20% or 25% or 30% or 35% or 40% or 45% of the protein present in the sample).
  • Any suitable protein purification method may be used as is standard practice in the art. Of course, it is typically solubilised as discussed above, and so the membrane protein is usually associated with detergent molecules and/or lipid molecules.
  • the first and second populations of the membrane protein are expressed and labelled separately.
  • the populations may also be solubilised and/or purified separately as discussed above.
  • the first and second populations of the membrane protein are then mixed together to obtain a sample.
  • the first population and second population of the membrane protein are present in the sample in a 1:1 molar ratio.
  • a 1:1 molar ratio is typically approximated by a 1:1 volume ratio. The approximation is believed to be close since size differences between the two populations are expected to be small and the purification efficiency is thought to be the same.
  • a 1:1 volume ratio is achieved by solubilising an equal number of cells from a first population of cells expressing the membrane protein and a second population of cells expressing the membrane protein, purifying the membrane proteins from each population identically and eluting the purified protein in the same volume.
  • the eluates are then mixed in equal volumes, so for example 250 ⁇ l of the first population is mixed with 250 ⁇ l of the second population and so on.
  • one of the populations of membrane proteins in the sample will be labelled with a donor label (not an acceptor label) and the other of the populations of membrane proteins will be labelled with an acceptor label (not a donor label).
  • acceptor label' and 'acceptor label' we include the meaning of any pairs of labels, where activation of the donor label permits a distance-dependent interaction with the acceptor label, which interaction produces a detectable signal. In this way, the detectable signal generated by interaction of donor and acceptor pairs can be used as a readout of the level of aggregation between membrane proteins from the first and second populations, and therefore conformational stability.
  • the interaction between the donor label and acceptor label involves the transfer of energy from the donor label to the acceptor label.
  • resonance energy transfer relates to a method where a donor label is in a close vicinity to an acceptor label. This generates an energy flow from the donor to the acceptor leading to a detection scheme where a signal is monitored through the donor or acceptor.
  • a luminescence resonance energy transfer system where the donor dye can be a down or up converting label. The donor is excited and as a consequence of the proximity principle the acceptor label is excited by the donor compound and a signal is detected at the emission wavelength of the acceptor compound.
  • the resonance energy transfer can be realised as a signal generating method or a method where the signal is quenched.
  • any element can be used to quench signal such as a dye or metal.
  • the emission wavelength of the donor molecule is detected.
  • Typical metal chelating or complexing agents or ligands used in time-resolved fluorometry are 3-(2-thienoly)-1,1,1-trifluoroacetone, 3-benzoyl-1,1,1-trifluoroacetone, coproporphyrins, porphyrins, 3-naphthoyl-1,1,1-trifluoroacetone, 2,2-dimethyl-4-perfluorobutyoyl-3-butanone, 2,2'-dipyridyl, phenanthroline, salicylic acid, phenanthroline carboxylic acid, aminophenanthroline, diphenylphenantroline, dimethylphenanthroline, bipyridylcarboxylic acid, aza crown ethers, trioctylphosphine oxide, aza cryptands, dibenzoylmethane, dinaphtoylmethane, dibiphenoylmethane, benzoylacetonato, phen
  • Metal ions can be, for example, lanthanide or ruthenium ions.
  • the interaction between the donor and acceptor labels involves fluorescence energy transfer (FRET), a phenomenon whereby when donor and acceptor fluorophores are in close proximity, the fluorescence energy of the donor fluorophore transfers to the acceptor fluorophore, and luminescence of the acceptor fluorophore is observed.
  • FRET fluorescence energy transfer
  • Any suitable pair of donor and acceptor fluorophores may be used as the donor and acceptor labels.
  • a particularly preferred donor fluorophore is a lanthanide, such as europium, terbium, samarium or dysprosium.
  • Lanthanides have long lifetimes that enable time-resolved measurement of acceptor emission. This is important because it significantly decreases the effect of autofluorescence from biological material and possible direct acceptor excitation. Also, the emission peaks of lanthanides are well separated, narrow and exhibit long Stokes' shifts (the distance between the maximum absorption and emission wavelengths).
  • the acceptor fluorophore may be any fluorophore with an excitation wave length that overlaps with one of the emission peaks of the donor.
  • the dominant peak is shown in bold.
  • any fluorophore with the excitation wave length around 490 nm, 545 nm or 620 nm could be used.
  • a fluorophore such as Fluorescein, Rhodamine, Alexa Fluora 488, Dylight 488, d2, Cy3, BODIPY FL, BODIPY 630/650-X, red-shifted variants of green fluorescent protein (GFP), typified by EGFP and GFP-S65T, may be used in combination with Terbium.
  • a fluorophore such as Fluorescein, Rhodamine, Alexa Fluora 488, Dylight 488, d2, Cy3, BODIPY FL, BODIPY 630/650-X, red-shifted variants of green fluorescent protein (GFP), typified by EGFP and GFP-S65T, may be used in combination with Terbium.
  • the interaction between the donor label and the acceptor label involves bioluminescence energy transfer (BRET).
  • BRET bioluminescence energy transfer
  • a criteria which should be considered in determining suitable pairings for BRET is the relative emission/fluorescence spectrum of the acceptor label compared to that of the bioluminescent protein.
  • the emission spectrum of the bioluminescent protein should overlap with the absorbance spectrum of the acceptor protein such that the light energy from the bioluminescent protein luminescence emission is at a wavelength that is able to excite the acceptor label and thereby promote acceptor label fluorescence when the two molecules are in a proper proximity and orientation with respect to one another.
  • BRET Renilla luciferase
  • RLuc Renilla luciferase
  • BRET 1 coelenterazine h
  • Clz400a coelenterazine 400a
  • BRET 2 ⁇ 395 nm
  • any suitable pairs of BRET donor and acceptor labels may be used that are known in the art, and as reviewed for example in Xia and Rao (Curr Opin Biol 20: 1-8, 2009 ).
  • Renilla luciferase/EGFP pairing has been compared to Renilla luciferase/EYEF pairing based on observable emission spectral peaks (Xu, 1999; Wang, et al (1997) in Bioluminescence and Chemiluminescence: Molecular Reporting with Photons, eds. Hastings et al (Wiley, New York), pp. 419-422 ).
  • protein fusions are prepared containing the selected bioluminescent protein and acceptor molecule and are tested, in the presence of an appropriate substrate.
  • the donor and acceptor labels are attached to the membrane protein at either the N-terminus or the C-terminus of the membrane protein. Labels attached to the termini are likely to have minimal effects on the structure and activity of the membrane protein. However, in principle, the labels can be placed in other regions of the membrane protein provided that they do not prevent folding of the membrane protein and/or interfere with its activity. Techniques to assess protein folding and activity to enable the skilled person to assess this are standard practice in the art.
  • the membrane proteins of the first and second populations are labelled with the donor and acceptor labels such that the labels are covalently attached to the membrane protein, for example at the N- or C- terminus of the membrane protein.
  • the membrane protein in the first population may be covalently attached to a donor label and the membrane protein in the second population may be covalently attached to an acceptor label, or the membrane protein in the first population may be covalently attached to an acceptor label and the membrane protein in the second population may be covalently attached to a donor label.
  • the Tag-liteTM system marketed by Cisbio may be used [7, 8, 9].
  • the membrane protein of interest is cloned into an expression vector upstream or downstream of a SNAP or a CLIP tag.
  • the SNAP and CLIP tags are related proteins that are modified forms of the mammalian O6-alkylguanine-DNA-alkyltransferase (AGT).
  • AGT mammalian O6-alkylguanine-DNA-alkyltransferase
  • the SNAP-tag and CLIP-tag substrates are derivates of benzyl purines and benzyl pyrimidines, where the benzyl group is attached to a functional group such as a fluorophore.
  • the modified benzyl group reacts with a free cysteine in AGT and the functional group is attached covalently.
  • the expression of the membrane protein from this plasmid results in the production of a membrane protein having the SNAP or CLIP tag at the N-terminus or C-terminus.
  • Membrane proteins can then be labeled with lanthanides (such as Terbium or Europium) that act as donor fluorophores.
  • the membrane protein may be labelled with a donor or acceptor label (eg a donor or acceptor fluorophore) via a terminus tag, eg a C-terminal or N-terminal tag, such as the SNAP and CLIP tags using the Tag-liteTM system.
  • a donor or acceptor label eg a donor or acceptor fluorophore
  • a terminus tag eg a C-terminal or N-terminal tag, such as the SNAP and CLIP tags using the Tag-liteTM system.
  • fusion polypeptides For donor and acceptor labels that are polypeptides (eg GFP), it will be understood that the labels may be covalently attached to the membrane protein by forming a fusion polypeptide. Preparation of fusion polypeptides is routine practice in the art and may involve chemical conjugation or may involve recombinant technology where the labelled membrane protein is expressed by a single polynucleotide.
  • the donor and acceptor labels may be attached to the membrane proteins non-covalently provided that the non-covalent interaction is strong enough to withstand the stability modulating agent and/or condition in step (b).
  • Such non-covalent bindings may include immunological bindings or bindings such as via biotin/avidin or streptavidin. Whether the non-covalent interaction is sufficiently strong can be determined by exposing the labelled membrane protein to the stability modulating agent and/or condition and using an appropriate analytical technique to assess whether the label remains bound to the membrane protein following exposure.
  • the donor label or acceptor label may be attached to the membrane directly or indirectly.
  • the membrane protein directly' we include the meaning of the label being either covalently or non-covalently attached to the membrane protein.
  • the membrane protein indirectly' we include the meaning of the label being either covalently or non-covalently attached to a further moiety which, in turn, is covalently or non-covalently attached to the membrane protein.
  • the donor label is activated to permit a distance-dependent interaction with the acceptor label, and the level of aggregation assessed.
  • activating the donor label we include the meaning of initiating a process whereby there is a distance-dependent interaction between the donor and acceptor labels.
  • the process may be one that results in the emission by the donor of a photon, or transfer of energy through resonance energy transfer or some other method.
  • donor activators include a photon (as in the case of a FRET donor) or a substrate of a bioluminescent protein (as in the case of a BRET donor).
  • activating the donor label may comprise irradiating the donor label or exposing the donor label to a substance, so as to lead to a distance dependent interaction with the acceptor label.
  • step (b) it may be desirable to also perform steps (a) and (c) of the method without performing step (b). This would provide an indication of background aggregation when the membrane protein is not exposed to a stability modulating agent and/or condition that can be compared to the level of aggregation when the membrane protein is exposed to a stability modulating agent and/or condition.
  • the assay may be preceded by a step wherein the aggregation between membrane proteins of the first and second populations is assessed without exposure to a stability modulating agent and/or condition.
  • a second aspect of the invention provides an assay for assessing the conformational stability of a membrane protein, comprising:
  • aggregation of the membrane protein is detected by virtue of the inability of the donor labels and acceptor labels to label the termini of the membrane protein when aggregated.
  • the membrane protein aggregates the labelling sites on the membrane protein become inaccessible and so the donor and acceptor labels cannot come into proximity with each other, and thereby cannot generate a detectable signal. If the membrane proteins are stable, there is less aggregation upon exposure to a destabilising agent and/or condition and so more signal, whereas if the membrane proteins are unstable, there is more aggregation upon exposure to a destabilising agent and/or condition and so less signal.
  • Preferences for the membrane protein population, donor label and acceptor label include those described above in relation to the first aspect of the invention.
  • one of the N-terminus or C-terminus of the membrane protein is capable of being labelled non-covalently with a donor label
  • the other of the N-terminus or C-terminus of the membrane protein is capable of being labelled non-covalently with an acceptor label
  • the membrane protein may be engineered to contain a moiety at the N- and/or C-terminus that is capable of, either directly or indirectly, binding to the donor or acceptor label non-covalently.
  • the N-terminus and/or C-terminus may contain a tag, such as a c-Myc tag, recognised by an antibody that, in turn, is labelled with the donor label or acceptor label.
  • the N-terminus and/or C-terminus may contain a moiety that is recognised by one member of a binding partner pair (eg biotin and streptavidin/avidin), the other member of the binding partner pair, in turn, being labelled with the donor label or acceptor label.
  • a binding partner pair eg biotin and streptavidin/avidin
  • Example 1 illustrates this principle whereby a GPCR is N-terminally tagged with a biotin acceptor peptide (BAP) and is C-terminally tagged with a c-Myc tag. The GPCR may then be labelled by mixing with streptavidin labelled with d2 and anti-c-Myc antibody labelled with terbium.
  • the labels do not need to be capable of recognising the tertiary structure of the membrane protein, and so a loss of labelling is a direct measure of protein aggregation that is not skewed by changes in ligand or antibody binding sites that are not necessarily correlated with aggregation.
  • one of the termini be labelled prior to step (b), provided that the labelling can withstand the stability modulating agent and/or condition.
  • the denaturant/denaturing condition is removed before labelling of one or both of the termini, so as to minimise adversely affecting the labelling procedure, as described further below.
  • the termini of the membrane protein are labelled. Either the N-terminus is labelled with a donor label and the C-terminus with an acceptor label, or the C-terminus is labelled with a donor label and the N-terminus with a donor label.
  • the amount of label that is added to the membrane protein is an amount that gives the maximal distance-dependent interaction signal while using the least amount of labelling reagents.
  • the amount can be readily determined empirically and will generally depend on the choice of label and expression levels of membrane protein. Optimising the amount may involve doing a titration matrix of varying amounts of acceptor and donor labels, on a lysate containing the membrane protein of interest as well as a mock lysate.
  • the optimal amount is one that gives the greatest difference in distance-dependent interaction signal (eg FRET signal) as between the lysate containing the membrane protein, and the mock lysate, while using the least amount of labelling reagents.
  • the inventors have performed such an optimisation for streptavidin-d2 and Myc-Tb, and identified respective optimum concentrations of 1 nM and 100 nM.
  • the level of aggregation is then assessed.
  • step (b) it may be desirable to also perform steps (a) and (c) of the method without performing step (b). This would provide an indication of background aggregation when the membrane protein is not exposed to a stability modulating agent and/or condition that can be compared to the level of aggregation when the membrane protein is exposed to a stability modulating agent and/or condition.
  • the assay may be preceded by a step wherein the aggregation between membrane proteins of the first and second populations is assessed without exposure to a stability modulating agent and/or condition.
  • the invention also provides a membrane protein (eg GPCR) wherein one of the N-terminus or C-terminus is attached to a donor label (eg donor fluorophore) and the other of the N-terminus or C-terminus is attached to an acceptor label (eg acceptor fluorophore).
  • a donor label eg donor fluorophore
  • acceptor label eg acceptor fluorophore
  • the membrane protein may be a GPCR that has the donor fluorophore (eg terbium) attached to the N-terminus and a donor fluorophore (eg EGFP) attached to the C-terminus.
  • the attachment of the labels to the termini of the membrane protein need not be direct, but may be indirect, eg by virtue of the label being attached to a further moiety, such as a terminal tag, which in turn is attached to the membrane protein.
  • the labels may be attached non-covalently or covalently to the termini of the membrane protein.
  • the invention also provides the use of such a membrane protein in an assay for measuring the conformational stability of the membrane protein.
  • the stability modulating agent and/or condition in the assays of the invention is a denaturing agent and/or condition, for example one selected from one or more of heat, pH, a detergent, or a chaotropic agent. It will be appreciated that any denaturing agent and/or condition may be used which is known to modulate the secondary and tertiary structure of a protein but not the primary structure of a protein.
  • thermostability In relation to stability to heat (ie thermostability), it may be convenient to determine a "quasi T m " or "quasi T agg " ie the temperature at which 50% of the membrane protein (eg GPCR) is aggregated under stated conditions after incubation for a given period of time (eg 30 minutes). Mutant membrane proteins that have higher thermostability have an increased quasi Tm or Tagg compared to their parents.
  • the membrane protein In relation to stability to a detergent or to a chaotrope, typically the membrane protein is incubated for a defined time in the presence of a test detergent or a test chaotropic agent and the stability is determined using an assay of the invention.
  • a typical test pH would be chosen (eg in the range 4.5 to 5.5 (low pH) or in the range 8.5 to 9.5 (high pH).
  • membrane proteins are stable in the presence of such detergents.
  • the order of "harshness" of certain detergents is DDM, C 11 ⁇ C 10 ⁇ C 9 ⁇ C 8 maltoside or glucoside, lauryldimethylamine oxide (LDAO) and SDS. It is particularly preferred if the membrane protein (eg GPCR) is more stable to any of C 9 maltoside or glucoside, C 8 maltoside or glucoside, LDAO and SDS, and so it is preferred that these detergents are used for stability testing.
  • thermostability may also be a guide to the stability to other denaturing or destabilising agents and/or conditions.
  • increased thermostability is likely to translate into stability in destabilising detergents, especially those that are more destabilising than DDM, eg those detergents with a smaller head group and a shorter alkyl chain and/or with a charged head group.
  • a thermostable GPCR is also more stable towards harsh detergents, for example.
  • the destabilising condition When an extreme of pH is used as the destabilising condition, it will be appreciated that this can be removed quickly by adding a neutralising agent.
  • a chaotrope when used as a destabilizing agent, the destabilising effect can be removed by diluting the sample below the concentration in which the chaotrope exerts its chaotropic effect.
  • a considerable advantage of the assays of the invention is that they do not need to include a separation step to remove labelled ligand. This is an important difference from known methods of measuring membrane protein stability which involve use of a radiolabelled ligand. In such known methods, unbound radiolabel must be removed in a washing step prior to detection of the radiolabel. This separation step makes the known methods laborious and unsuitable for high throughput applications.
  • the assays of the invention do not use ligand binding as an indicator of the stability of a membrane protein, it may be beneficial to include a ligand of the membrane protein in the sample provided in step (a).
  • a ligand of the membrane protein in the sample provided in step (a).
  • the presence of a ligand, especially when the membrane protein is a GPCR, can improve stability and/or increase the probability of stabilising the GPCR in a desired conformation.
  • particular conformational states may be enriched for by varying physiochemical parameters or using additives such as any of pH, salt, metal ions, temperature, chaotropic agents, and glycerol.
  • One may optimise physiochemical parameters to favour a particular conformation by doing standard pharmacological characterisations such as ligand binding affinities or functional assays under different parameters.
  • ligand we include any molecule which binds to a membrane protein such as a GPCR.
  • the ligand When the membrane protein is a GPCR, the ligand will typically bind to one conformation of the GPCR (and may cause the GPCR to adopt this conformation), but does not bind as strongly to another conformation that the GPCR may be able to adopt. Thus, the presence of the ligand may be considered to encourage the GPCR to adopt the particular conformation.
  • the ligand is from the agonist class of ligands and the particular conformation is an agonist conformation, or the ligand is from the antagonist class of ligands and the particular conformation is an antagonist conformation.
  • the ligand is a full agonist and is able to bind to the membrane protein (eg a receptor such as a GPCR) and is capable of eliciting a full (100%) biological response, measured for example by G-protein coupling, downstream signalling events or a physiological output such as vasodilation.
  • the biological response is GDP/GTP exchange in a G-protein, followed by stimulation of the linked effector pathway.
  • the measurement typically, is GDP/GTP exchange or a change in the level of the end product of the pathway (eg cAMP, cGMP or inositol phosphates).
  • the ligand may also be a partial agonist and is able to bind to the GPCR and is capable of eliciting a partial ( ⁇ 100%) biological response.
  • the ligand may also be an inverse agonist, which is a molecule which binds to a membrane protein (eg a receptor such as a GPCR) and reduces its basal (ie unstimulated by agonist) activity sometimes even to zero.
  • a membrane protein eg a receptor such as a GPCR
  • the ligand may also be an antagonist, which is a molecule which binds to a membrane protein (eg a receptor such as a GPCR) and blocks binding of an agonist, so preventing a biological response.
  • a membrane protein eg a receptor such as a GPCR
  • Inverse agonists and partial agonists may under certain assay conditions be antagonists.
  • the above ligands may be orthosteric, by which we include the meaning that they combine with the same site as the endogenous agonist; or they may be allosteric or allotopic, by which we include the meaning that they combine with a site distinct from the orthosteric site.
  • the above ligands may be syntopic, by which we include the meaning that they interact with other ligand(s) at the same or an overlapping site. They may be reversible or irreversible.
  • antagonists In relation to antagonists, they may be surmountable, by which we include the meaning that the maximum effect of agonist is not reduced by either pre-treatment or simultaneous treatment with antagonist; or they may be insurmountable, by which we include the meaning that the maximum effect of agonist is reduced by either pre-treatment or simultaneous treatment with antagonist; or they may be neutral, by which we include the meaning the antagonist is one without inverse agonist or partial agonist activity.
  • Antagonists typically are also inverse agonists.
  • Ligands for use in the invention may also be allosteric modulators such as positive allosteric modulators, potentiators, negative allosteric modulators and inhibitors. They may have activity as agonists or inverse agonists in their own right or they may only have activity in the presence of an agonist or inverse agonist in which case they are used in combination with such molecules in order to bind to the membrane protein (eg GPCR).
  • allosteric modulators such as positive allosteric modulators, potentiators, negative allosteric modulators and inhibitors. They may have activity as agonists or inverse agonists in their own right or they may only have activity in the presence of an agonist or inverse agonist in which case they are used in combination with such molecules in order to bind to the membrane protein (eg GPCR).
  • the above-mentioned ligands are small organic or inorganic moieties, but they may be peptides or polypeptides.
  • the ligand when it is a small organic or inorganic moiety, it has a M r of from 50 to 2000, such as from 100 to 1000, for example from 100 to 500.
  • the ligand binds to the membrane protein (eg GPCR) with a K d of from mM to pM, such as in the range of from ⁇ M (micromolar) to nM. Generally, the ligands with the lowest Kd are preferred.
  • the membrane protein eg GPCR
  • Small organic molecule ligands are well known in the art.
  • Other small molecule ligands include 5HT which is a full agonist at the 5HT1A receptor; eltoprazine which is a partial agonist at the 5HT1A receptor (see Newman-Tancredi et al (1997) Neurophamacology 36, 451-459 ); (+)-butaclamol and spiperone are dopamine D2 receptor inverse agonists (see Roberts & Strange (2005) Br. J. Pharmacol. 145, 34-42 ); and WIN55212-3 is a neutral antagonist of CB2 ( Savinainen et al (2005) Br. J. Pharmacol. 145, 636-645 ).
  • the ligand may be a peptidomimetic, a nucleic acid, a peptide nucleic acid (PNA) or an aptamer. It may be an ion such as Na + or Zn 2+ , a lipid such as oleamide, or a carbohydrate such as heparin.
  • the ligand may be a polypeptide which binds to the membrane protein (eg receptor such as a GPCR).
  • polypeptides by which we include oligopeptides
  • the polypeptide may be a naturally occurring GPCR-interacting protein or other protein which interacts with the GPCR, or a derivative or fragment thereof, provided that it binds selectively to the GPCR in a particular conformation.
  • GPCR-interacting proteins include those associated with signalling and those associated with trafficking, which often act via PDZ domains in the C terminal portion of the GPCR.
  • Polypeptides which are known to bind certain GPCRs include any of a G protein, an arrestin, a RGS protein, G protein receptor kinase, a RAMP, a 14-3-3 protein, a NSF, a periplakin, a spinophilin, a GPCR kinase, a receptor tyrosine kinase, an ion channel or subunit thereof, an ankyrin and a Shanks or Homer protein.
  • Other polypeptides include NMDA receptor subunits NR1 or NR2a, calcyon, or a fibronectin domain framework.
  • the polypeptide may be one which binds to an extracellular domain of a GPCR, such as fibulin-1.
  • the polypeptide may be another GPCR, which binds to the selected GPCR in a hetero-oligomer.
  • GPCRs A review of protein-protein interactions at GPCRs is found in Milligan & White (2001) Trends Pharmacol. Sci. 22, 513-518 , or in Bockaert et al (2004) Curr. Opinion Drug Discov. Dev. 7, 649-657 .
  • the polypeptide ligand may conveniently be an antibody which binds to the membrane protein (eg GPCR).
  • antibody we include naturally-occurring antibodies, monoclonal antibodies and fragments thereof. We also include engineered antibodies and molecules which are antibody-like in their binding characteristics, including single chain Fv (scFv) molecules and domain antibodies (dAbs). Mention is also made of camelid antibodies and engineered camelid antibodies. Such molecules which bind GPCRs are known in the art and in any event can be made using well known technology. Suitable antibodies include ones presently used in radioimmunoassay (RIAs) for GPCRs since they tend to recognise conformational epitopes.
  • RIAs radioimmunoassay
  • the polypeptide may also be a binding protein based on a modular framework, such as ankyrin repeat proteins, armadillo repeat proteins, leucine rich proteins, tetratriopeptide repeat proteins or Designed Ankyrin Repeat Proteins (DARPins) or proteins based on lipocalin or fibronectin domains or Affilin scaffolds based on either human gamma crystalline or human ubiquitin.
  • a modular framework such as ankyrin repeat proteins, armadillo repeat proteins, leucine rich proteins, tetratriopeptide repeat proteins or Designed Ankyrin Repeat Proteins (DARPins) or proteins based on lipocalin or fibronectin domains or Affilin scaffolds based on either human gamma crystalline or human ubiquitin.
  • antibodies or other "universal" binding polypeptides (such as G-proteins which are known to couple with many different GPCRs) may be particularly advantageous in the use of the method on "orphan" GPCRs for which the natural ligand, and small molecule ligands, are not known.
  • the above assays may be used in methods to select membrane proteins (eg GPCRs) with increased conformational stability.
  • Such methods have the advantage over existing methods that there is no need for a separation step to remove labelled ligand.
  • the method can be performed even where a good radioligand with high affinity and/or favourable properties in detergent is not available.
  • a third aspect of the invention provides a method for selecting a membrane protein (eg GPCR) with increased conformational stability, comprising:
  • the method of the third aspect of the invention may be considered to be a way of selecting membrane proteins (eg GPCRs) which are trapped in a conformation of biological relevance (eg ligand bound state), and which are more stable with respect to that conformation.
  • the ligand is from the agonist class of ligands and the particular conformation is an agonist conformation, or the ligand is from the antagonist class of ligands and the particular conformation is an antagonist conformation.
  • the mutant membrane proteins may be prepared by any suitable method.
  • the mutant protein is encoded by a suitable nucleic acid molecule and is expressed in a suitable host cell.
  • suitable nucleic acid molecules, expression systems and host cells include those described above.
  • the membrane protein is CCR5
  • the donor label is terbium
  • the acceptor label is EGFP.
  • Terbium may be attached to the N-terminus by using SNAP tag technology.
  • EGFP may be attached to the C-terminus by being expressed as a C-terminal fusion.
  • the membrane protein is CCR5
  • the donor label is terbium
  • the acceptor label is d2.
  • Terbium may be non-covalently attached to the C-terminus via labelling an anti-c-Myc antibody that is bound to a c-Myc epitope at the C-terminus.
  • d2 may be non-covalently attached to the N-terminus via labelling streptavidin that is bound to biotin which in turn binds biotin acceptor peptide (BAP) fused to the N-terminus.
  • the membrane protein is a GPCR such as any of GLP1R, CMKLR, or TGR5 (GPBAR-I), the donor label is terbium and the acceptor label is EGFP.
  • GPCR such as any of GLP1R, CMKLR, or TGR5 (GPBAR-I)
  • the donor label is terbium
  • the acceptor label is EGFP.
  • Terbium may be attached to the N-terminus by using SNAP tag technology.
  • EGFP may be attached to the C-terminus by being expressed as a C-terminal fusion.
  • the membrane protein is a GPCR such as any of GLP1R, CMKLR, or TGR5 (GPBAR-I)
  • the donor label is terbium and the acceptor label is d2.
  • Terbium may be non-covalently attached to the C-terminus via labelling an anti-c-Myc antibody that is bound to a c-Myc epitope at the C-terminus.
  • d2 may be non-covalently attached to the N-terminus via labelling streptavidin that is bound to biotin which in turn binds biotin acceptor peptide (BAP) fused to the N-terminus.
  • Protein aggregation upon unfolding is a general feature of all proteins that occurs to varying degrees in different proteins.
  • Membrane proteins are known to exhibit high levels of aggregation after solubilisation and more so after denaturation. It is therefore possible to use the levels of aggregation as a measure of global stability.
  • An assay that allows aggregation to be measured in an easy and miniaturisable format would be a useful tool to generate a stable receptor.
  • aggregation could be measured including the biophysical methods described above. Most of these methods require high amounts of very pure protein.
  • This assay is a variation on the previous method.
  • the FRET acceptor and donor are placed on the N- and C-termini of the same receptor molecule.
  • the receptor is solubilised and heated prior to labelling with the FRET acceptor and donor. So, as the receptor unfolds and aggregates the sites of FRET acceptor and donor labelling are obscured and thus become inaccessible. This in turn results in loss of FRET as a function of temperature and the T agg is defined as the temperature at which 50% FRET is observed ( Figure 2a ).
  • the receptor is tagged N-terminally with a biotin acceptor peptide (BAP) and is C-terminally tagged with cMyc tag.
  • the receptor is co-expressed with biotin ligase A (BirA) enzyme which leads to the expression of an N-terminally biotinylated receptor.
  • BirA biotin ligase A
  • the crude lysate is incubated at different temperatures for a set amount of time.
  • the lysates are then mixed with streptavidin labelled with d2 and anti-cMyc antibody labelled with terbium.
  • the levels of FRET are then measured and plotted against temperature ( Figure 2b ).
  • thermostabilisng mutations a number of mutants were generated in the plasmid encoding GLP1R using site directed mutagenesis. The intramolecular aggregation assay was then used to assess the thermal stability of the mutants compared to wild-type GLP1R.
  • Figure 3 shows exemplary data for T355F mutation that confers ⁇ 6°C increase in thermal stability of GLP1R. As seen from the figure, the value of Tagg for the stabilised mutant GLP1R is higher than that of the wild-type GLP1R.
  • the receptors were expressed transiently in HEK293T cells. Briefly, cells were seeded at the density of 3x10 6 cells in 10 cm petri dishes containing Dulbecco's Modified Eagle Medium (DMEM) supplemented with 10% foetal bovine serum (FBS) and incubated overnight in 37°C incubator. The next day, cells were transfected using 6ug of plasmid encoding the receptor of interest using GeneJuice according to the manufacturer's instructions. Cells were incubated for about 40 hours post-transfection at the 37°C incubator.
  • DMEM Dulbecco's Modified Eagle Medium
  • FBS foetal bovine serum
  • solubilisation buffer containing 50 mM 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid (HEPES) and 150 mM sodium chloride adjusted to pH 7.5, supplemented with 1% n-Dodecyl- ⁇ -maltoside (DDM) and protease inhibitor cocktail. Receptors were solubilised for 1 hour with end-to-end rotation. All solubilisation steps as well as the subsequent steps were carried out at 4°C.
  • HEPES 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid
  • DDM n-Dodecyl- ⁇ -maltoside
  • DDM n-Dodecyl- ⁇ -maltoside
  • SNAP-tagged and EGFP-tagged receptors were expressed separately in HEK293T cells as described above. Prior to solubilisation, SNAP-tagged receptors were harvested and re-suspended in DMEM plus FBS containing 250 nM of SNAP-Lumi4Tb (Cisbio) and incubated for 1 hour at 37°C incubator to label the N-terminus. Cells were then washed three times with 1mL of PBS to remove the excess unbound SNAP-Lumi4Tb. After the last wash cells were solubilised in parallel along with cells expressing EGFP tagged receptor in 1% DDM. The crude lysates were clarified by centrifugation at 13000 rpm for 10 minutes at 4°C.
  • the cleared lysates were incubated with 250 uL of 50% slurry Ni-NTA agarose resin pre-equilibrated in the solubilisation buffer in order to purify receptors using the C-terminal 10x His tag.
  • the mixture was incubated at 4°C with end-to-end rotation for 1.5 hour and then washed 3x with 1mL of chilled wash buffer containing 50mM HEPES, 150mM NaCl, 0.03%DDM, 20mM Imidazole, pH adjusted to 7.5.
  • After the last wash receptors were eluted in 500 uL of elution buffer containing 50mM HEPES, 150mM NaCl, 0.03% DDM and 100 mM Histidine, pH adjusted to 7.5.
  • the Lumi4Tb tagged and EGFP tagged samples were mixed 1:1 and aliquots were incubated at increasing temperatures for 30 minutes. The samples were then returned to 4°C and transferred to white 96-well plates and FRET was measured between Lumi4Tb and EGFP using PHERAstar Plus (BMG Labtech) instrument. The FRET settings were according to the recommendations of the manufacturer.
  • Receptors tagged N-terminally with the biotin acceptor tag (BAP) and C-terminally with c-Myc tag were expressed transiently in HEK293T cells as described above. It is notable that the plasmid also encodes for the BirA enzyme that mediates biotinylation on the BAP tag. Cells were incubated with 100uM of biotin during expression. Following harvesting and solubilisation, aliquots of cleared lysates were incubated at increasing temperatures for 30 minutes.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Immunology (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Molecular Biology (AREA)
  • Hematology (AREA)
  • Urology & Nephrology (AREA)
  • Biomedical Technology (AREA)
  • Physics & Mathematics (AREA)
  • General Health & Medical Sciences (AREA)
  • Biochemistry (AREA)
  • Cell Biology (AREA)
  • Medicinal Chemistry (AREA)
  • General Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • Pathology (AREA)
  • Food Science & Technology (AREA)
  • Microbiology (AREA)
  • Biotechnology (AREA)
  • Organic Chemistry (AREA)
  • Biophysics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Toxicology (AREA)
  • Genetics & Genomics (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Zoology (AREA)
  • Endocrinology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Bioinformatics & Computational Biology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Optics & Photonics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Investigating Or Analysing Biological Materials (AREA)
  • Peptides Or Proteins (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)

Claims (15)

  1. Test d'évaluation de la stabilité de conformation d'une protéine membranaire, comprenant :
    (a) la production d'un échantillon contenant une première population et une seconde population d'une protéine membranaire ; la protéine membranaire de la première population étant marquée avec un marqueur donneur et la protéine membranaire de la seconde population étant marquée avec un marqueur accepteur, ou la protéine membranaire de la première population étant marquée avec un marqueur accepteur et la protéine membranaire de la seconde population étant marquée avec un marqueur donneur, et les populations de protéine membranaire étant produites sous une forme solubilisée dans laquelle les protéines membranaires conservent leur intégrité structurelle et se présentent sous une forme fonctionnelle,
    (b) l'exposition des première et seconde populations de la protéine membranaire à un dénaturant ou une condition dénaturante,
    (c) et l'évaluation de l'agrégation entre les protéines membranaires des première et seconde populations par activation du marqueur donneur pour permettre une interaction, dépendant de la distance, avec le marqueur accepteur, laquelle interaction produit un signal détectable.
  2. Test selon la revendication 1, dans lequel la première population et la seconde population de la protéine membranaire sont présentes dans l'échantillon dans un rapport de 1:1.
  3. Test selon la revendication 1 ou 2, dans lequel le marqueur donneur est fixé de manière covalente à la protéine membranaire et le marqueur accepteur est fixé de manière covalente à la protéine membranaire.
  4. Test d'évaluation de la stabilité de conformation d'une protéine membranaire, comprenant :
    (a) la production d'un échantillon contenant une population de protéines membranaires, la population de protéines membranaires étant produite sous une forme solubilisée dans laquelle les protéines membranaires conservent une intégrité structurale et se présentent sous une forme fonctionnelle,
    (b) l'exposition de la population de protéines membranaires à un dénaturant ou une condition dénaturante,
    (c) le marquage de l'une des extrémités N-terminale ou C-terminale de la protéine membranaire avec un marqueur donneur et l'autre de l'extrémité N-terminale ou C-terminale de la protéine membranaire avec un marqueur accepteur,
    (d) et l'évaluation de l'agrégation des protéines membranaires dans la population par activation du marqueur donneur pour permettre une interaction, dépendant de la distance, avec le marqueur accepteur, laquelle interaction produit un signal détectable.
  5. Test selon l'une quelconque des revendications 1 à 4, dans lequel l'interaction entre le marqueur donneur et le marqueur accepteur implique le transfert d'énergie d'un fluorophore donneur à un fluorophore accepteur, facultativement le fluorophore donneur étant un lanthanide, tel que le Terbium ; et/ou le fluorophore accepteur étant EGFP ou d2.
  6. Test selon l'une quelconque des revendications 1 à 5, dans lequel l'interaction entre le marqueur donneur et le marqueur accepteur est une réaction chimioluminescente, facultativement dans lequel l'interaction entre le marqueur donneur et le marqueur accepteur implique la génération de molécules d'oxygène singulet qui déclenchent une réaction chimioluminescente.
  7. Test selon l'une quelconque des revendications 1 à 6, dans lequel le marqueur donneur et/ou le marqueur accepteur est lié directement à la protéine membranaire, ou dans lequel le marqueur donneur et/ou le marqueur accepteur est lié indirectement à la protéine membranaire.
  8. Test selon l'une quelconque des revendications 1 à 7, dans lequel l'échantillon produit à l'étape (a) contient un ou plusieurs détergents choisis parmi le dodécylmaltoside (DDM) ; le maltoside ou glucoside en C11, C10, C9 ou C8 ; l'oxyde de lauryldiméthylamine (LDAO) ; et le dodécylsulfate de sodium (SDS).
  9. Test selon l'une quelconque des revendications 1 à 8, dans lequel le dénaturant ou la condition dénaturante est choisi parmi un ou plusieurs parmi la chaleur, un détergent, un agent chaotropique ou le pH.
  10. Test selon l'une quelconque des revendications 1 à 9, dans lequel la protéine membranaire est un récepteur couplé à la protéine G (GPCR), facultativement dans lequel l'échantillon produit à l'étape (a) contient un ligand GPCR, le ligand étant un ligand qui se lie à un GPCR lorsque le GPCR se trouve dans une conformation particulière, facultativement dans lequel le ligand GPCR provient de la classe agoniste des ligands et la conformation particulière est une conformation agoniste, ou le ligand GPCR provient de la classe antagoniste des ligands et la conformation particulière est une conformation antagoniste.
  11. Test selon l'une quelconque des revendications précédentes, le test étant utilisé dans le criblage de médicaments.
  12. Procédé de sélection d'une protéine membranaire ayant une stabilité de conformation majorée, ledit test comprenant :
    (a) la comparaison de la stabilité de conformation d'un ou de plusieurs mutants d'une protéine membranaire parente avec la stabilité de conformation de la protéine membranaire parente conformément au test selon l'une quelconque des revendications 1 à 11, et
    (b) la sélection d'un ou de plusieurs mutants qui ont une stabilité de conformation majorée par rapport à la protéine membranaire parente, facultativement la protéine membranaire étant un GPCR.
  13. Procédé selon la revendication 12, comprenant :
    (a) la production d'un ou de plusieurs mutants d'une protéine membranaire parente ;
    (b) l'évaluation de la stabilité de conformation du ou des mutants de la protéine membranaire parente conformément au test selon l'un quelconque des principes 1 à 11 ;
    (c) l'évaluation de la stabilité de conformation de la protéine membranaire parente conformément au test selon l'une quelconque des revendications 1 à 11 ; et
    (d) la sélection d'un ou de plusieurs mutants de la protéine membranaire parente ayant une stabilité de conformation majorée par rapport à la stabilité de conformation de la protéine parente.
  14. Procédé selon la revendication 12 ou 13, dans lequel la protéine membranaire a une stabilité majorée par rapport à l'un quelconque parmi la chaleur, un détergent, un agent chaotropique ou un extrême de pH.
  15. Procédé de préparation d'un GPCR mutant, le procédé comprenant :
    (a) la mise en oeuvre du procédé selon l'une quelconque des revendications 12 à 14,
    (b) l'identification de la position ou des positions du résidu ou des résidus aminoacides mutés dans la protéine membranaire mutante ou les protéines membranaires mutantes qui ont été sélectionnées pour la stabilité majorée, et
    (c) la synthèse d'une protéine membranaire mutante qui contient un acide aminé de remplacement à une ou plusieurs des positions identifiées.
EP13734141.8A 2012-06-01 2013-05-31 Essais Active EP2856161B1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201261654265P 2012-06-01 2012-06-01
PCT/GB2013/051464 WO2013179062A2 (fr) 2012-06-01 2013-05-31 Essais

Publications (2)

Publication Number Publication Date
EP2856161A2 EP2856161A2 (fr) 2015-04-08
EP2856161B1 true EP2856161B1 (fr) 2018-04-18

Family

ID=48746572

Family Applications (1)

Application Number Title Priority Date Filing Date
EP13734141.8A Active EP2856161B1 (fr) 2012-06-01 2013-05-31 Essais

Country Status (7)

Country Link
US (1) US10458993B2 (fr)
EP (1) EP2856161B1 (fr)
JP (1) JP6251252B2 (fr)
CN (1) CN104350383B (fr)
DK (1) DK2856161T3 (fr)
ES (1) ES2676827T3 (fr)
WO (1) WO2013179062A2 (fr)

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8785135B2 (en) 2007-03-22 2014-07-22 Heptares Therapeutics Limited Mutant G-protein coupled receptors and methods for selecting them
GB0724860D0 (en) 2007-12-20 2008-01-30 Heptares Therapeutics Ltd Screening
WO2013021206A2 (fr) 2011-08-10 2013-02-14 Heptares Therapeutics Limited Protéines stables
JP6473366B2 (ja) * 2015-03-31 2019-02-20 シスメックス株式会社 温度判定方法、標的ペプチドの検出方法および温度判定試薬
KR101862198B1 (ko) * 2016-09-30 2018-05-30 (주)스파크바이오파마 2차원 겔 전기영동에서의 열 안정성 변화-기반 형광 차이를 이용한 표적 단백질의 규명 방법

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020048811A1 (en) * 2000-10-16 2002-04-25 Devreotes Peter N. Receptor mediated activation of heterotrimeric G-proteins
US20090220988A1 (en) * 2005-08-30 2009-09-03 Eric Trinquet Method of revealing a biological process using a fret measurement
WO2011143575A2 (fr) * 2010-05-13 2011-11-17 Cornell University Smfret avec des protéines membranaires
EP2450445A1 (fr) * 2007-03-22 2012-05-09 Heptares Therapeutics Limited Récepteur couplé aux protéines G mutant avec une stabilité conformationelle élevée

Family Cites Families (27)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2327400A1 (fr) 1998-04-06 1999-10-14 Bunsen Rush Laboratories, Inc. Biocapteurs par evolution dirigee
US6692696B1 (en) 1998-06-18 2004-02-17 ARETé ASSOCIATES Biosensor
WO2000070343A2 (fr) 1999-05-14 2000-11-23 Repliscent, Inc. Biocapteurs et systemes de replication sens bases sur des recepteurs couples a la proteine g (gprc)
US20020061834A1 (en) 2000-02-09 2002-05-23 Rosen Craig A. Human G-protein Chemokine receptor (CCR5) HDGNR10
US7678539B2 (en) 2000-08-10 2010-03-16 Corning Incorporated Arrays of biological membranes and methods and use thereof
JP2005503124A (ja) 2001-02-14 2005-02-03 アムジェン インコーポレイテッド Gタンパク質共役受容体分子及びその使用
US7294472B2 (en) 2001-03-14 2007-11-13 Caden Biosciences Method for identifying modulators of G protein coupled receptor signaling
US20050009204A1 (en) 2003-07-11 2005-01-13 Ye Fang Multiplexed binding assays for receptor arrays
JP2004238384A (ja) 2002-03-28 2004-08-26 Takeda Chem Ind Ltd 新規スクリーニング方法
AU2003227268A1 (en) 2002-03-28 2003-10-13 Takeda Chemical Industries, Ltd. Novel screening method
US20040096914A1 (en) 2002-11-20 2004-05-20 Ye Fang Substrates with stable surface chemistry for biological membrane arrays and methods for fabricating thereof
US7803559B1 (en) 2003-08-08 2010-09-28 The Regents Of The University Of Ca Protein aggregation regulators
JP2006340717A (ja) 2005-05-11 2006-12-21 Sekisui Chem Co Ltd Gタンパク質共役型受容体に対する結合性評価方法及び評価用組成物、融合タンパク質、並びに、遺伝子
AU2008311869B2 (en) 2007-10-17 2013-11-21 The Board Of Trustees Of The Leland Stanford Junior University Method and composition for crystallizing G protein-coupled receptors
JP5560448B2 (ja) 2007-10-22 2014-07-30 ザ スクリプス リサーチ インスティテュート 膜タンパク質の高分解能結晶を得るための方法および組成物
GB0724051D0 (en) 2007-12-08 2008-01-16 Medical Res Council Mutant proteins and methods for producing them
GB0724860D0 (en) 2007-12-20 2008-01-30 Heptares Therapeutics Ltd Screening
GB0802474D0 (en) 2008-02-11 2008-03-19 Heptares Therapeutics Ltd Mutant proteins and methods for selecting them
WO2008068534A2 (fr) 2008-03-05 2008-06-12 Heptares Therapeutics Limited Structure cristalline
GB0910725D0 (en) 2009-06-22 2009-08-05 Heptares Therapeutics Ltd Mutant proteins and methods for producing them
US8900591B2 (en) 2010-08-20 2014-12-02 Heptares Therapeutics Limited Vaccines comprising mutant GPCRs with increased conformational stability relative to parent receptors
WO2012030735A1 (fr) 2010-08-30 2012-03-08 Confometrx, Inc. Procédé et composition de cristallisation de rcpg de famille c
WO2012098413A1 (fr) 2011-01-21 2012-07-26 Heptares Therapeutics Limited Protéines mutantes de récepteur couplé à une protéine g et leurs procédés de production
EA201391514A1 (ru) 2011-05-13 2014-08-29 Рецептос, Инк Новые партнеры слияния для кристаллизации рецепторов, связанных с g-белками
WO2013021206A2 (fr) 2011-08-10 2013-02-14 Heptares Therapeutics Limited Protéines stables
EP2617732A1 (fr) * 2012-01-19 2013-07-24 Vib Vzw Outils et procédés pour l'expression de protéines de membrane
US20150261911A1 (en) 2014-03-13 2015-09-17 Heptares Therapeutics Limited Crystal structure

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020048811A1 (en) * 2000-10-16 2002-04-25 Devreotes Peter N. Receptor mediated activation of heterotrimeric G-proteins
US20090220988A1 (en) * 2005-08-30 2009-09-03 Eric Trinquet Method of revealing a biological process using a fret measurement
EP2450445A1 (fr) * 2007-03-22 2012-05-09 Heptares Therapeutics Limited Récepteur couplé aux protéines G mutant avec une stabilité conformationelle élevée
WO2011143575A2 (fr) * 2010-05-13 2011-11-17 Cornell University Smfret avec des protéines membranaires

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
ADAM M. KNEPP ET AL: "Direct Measurement of Thermal Stability of Expressed CCR5 and Stabilization by Small Molecule Ligands", BIOCHEMISTRY, vol. 50, no. 4, 1 February 2011 (2011-02-01), pages 502 - 511, XP055078620, ISSN: 0006-2960, DOI: 10.1021/bi101059w *
ANBAZHAGAN V ET AL: "Unfolding a transmembrane helix dimer: A FRET study in mixed micelles", ARCHIVES OF BIOCHEMISTRY AND BIOPHYSICS, ACADEMIC PRESS, US, vol. 495, no. 2, 15 March 2010 (2010-03-15), pages 159 - 164, XP026924525, ISSN: 0003-9861, [retrieved on 20100113], DOI: 10.1016/J.ABB.2010.01.006 *
JUNPING FAN ET AL: "Thermal precipitation fluorescence assay for protein stability screening", JOURNAL OF STRUCTURAL BIOLOGY, ACADEMIC PRESS, UNITED STATES, vol. 175, no. 3, 5 May 2011 (2011-05-05), pages 465 - 468, XP028276636, ISSN: 1047-8477, [retrieved on 20110512], DOI: 10.1016/J.JSB.2011.05.003 *

Also Published As

Publication number Publication date
ES2676827T3 (es) 2018-07-25
DK2856161T3 (en) 2018-07-23
WO2013179062A3 (fr) 2014-03-06
JP6251252B2 (ja) 2017-12-20
CN104350383A (zh) 2015-02-11
CN104350383B (zh) 2017-03-08
JP2015519569A (ja) 2015-07-09
WO2013179062A2 (fr) 2013-12-05
US20150147822A1 (en) 2015-05-28
US10458993B2 (en) 2019-10-29
EP2856161A2 (fr) 2015-04-08

Similar Documents

Publication Publication Date Title
EP2856161B1 (fr) Essais
JP4955391B2 (ja) Gタンパク質共役受容体およびそのシグナル伝達経路の断片相補アッセイ
US20130035256A1 (en) Chimeric Polypeptides Useful in Proximal and Dynamic High-Throughput Screening Methods
US11041868B2 (en) Compositions comprising G protein-coupled receptors maintained in a conformationally selective manner by a modulator
Kim et al. Cellular and molecular biology of orphan G protein‐coupled receptors
US20150024410A1 (en) Method for determining the glycosylation of an antibody
US8697372B2 (en) Method for determining the binding of a given compound to a membrane receptor
KR20100058526A (ko) 프로테아제 활성화된 리포터를 사용한 단백질-단백질 상호작용 조절 분자의 확인 방법
Dal Maso et al. Characterization of signalling and regulation of common calcitonin receptor splice variants and polymorphisms
Milligan et al. G protein-coupled receptor fusion proteins in drug discovery
Héroux et al. Assembly and signaling of CRLR and RAMP1 complexes assessed by BRET
A Kaczor et al. Application of BRET for studying G protein-coupled receptors
CN111164427B (zh) 用于测量对g蛋白偶联受体活性的调控的方法
Daly et al. β-Arrestin-dependent and-independent endosomal G protein activation by the vasopressin type 2 receptor
Böhme et al. Tracking of human Y receptors in living cells—a fluorescence approach
US20080274913A1 (en) Multiplex Array Useful for Assaying Protein-Protein Interaction
Okamoto et al. Human chorionic gonadotropin (hCG) beta-core fragment is produced by degradation of hCG or free hCG beta in gestational trophoblastic tumors: a possible marker for early detection of persistent postmolar gestational trophoblastic disease
US20230091315A1 (en) Anti-g-protein alpha antibody
JP2008518600A (ja) Gタンパク質共役型レセプターおよびそのリガンドをアッセイするための組成物および方法
EP2329268B1 (fr) Méthodes et composés pour un essai de liaison d'un ligand à un récepteur couplé à la protéine g
US20110244487A1 (en) Methods for testing binding of a ligand to a g protein-coupled receptor
Dijon Novel complementation biosensors to investigate G protein-coupled receptor kinetics and signalling bias
US20120021440A1 (en) Methods for testing ligand binding to g protein-coupled receptors
Toth et al. ArreSTick Motif is Responsible for GPCR-beta-Arrestin Binding Stability and Extends Phosphorylation-Dependent beta-arrestin Interactions to Non-Receptor Proteins
Adamczuk et al. Glycoprotein-glycoprotein receptor binding detection using bioluminescence resonance energy transfer (BRET)

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20141222

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

AX Request for extension of the european patent

Extension state: BA ME

DAX Request for extension of the european patent (deleted)
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

17Q First examination report despatched

Effective date: 20170314

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: GRANT OF PATENT IS INTENDED

INTG Intention to grant announced

Effective date: 20171030

GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE PATENT HAS BEEN GRANTED

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

REG Reference to a national code

Ref country code: GB

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: CH

Ref legal event code: EP

REG Reference to a national code

Ref country code: AT

Ref legal event code: REF

Ref document number: 991041

Country of ref document: AT

Kind code of ref document: T

Effective date: 20180515

REG Reference to a national code

Ref country code: IE

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 6

REG Reference to a national code

Ref country code: DE

Ref legal event code: R096

Ref document number: 602013036079

Country of ref document: DE

REG Reference to a national code

Ref country code: DK

Ref legal event code: T3

Effective date: 20180716

REG Reference to a national code

Ref country code: SE

Ref legal event code: TRGR

REG Reference to a national code

Ref country code: ES

Ref legal event code: FG2A

Ref document number: 2676827

Country of ref document: ES

Kind code of ref document: T3

Effective date: 20180725

REG Reference to a national code

Ref country code: NL

Ref legal event code: MP

Effective date: 20180418

REG Reference to a national code

Ref country code: LT

Ref legal event code: MG4D

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: AL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180418

Ref country code: BG

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180718

Ref country code: NO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180718

Ref country code: LT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180418

Ref country code: FI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180418

Ref country code: PL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180418

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: HR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180418

Ref country code: GR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180719

Ref country code: LV

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180418

Ref country code: RS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180418

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: PT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180820

REG Reference to a national code

Ref country code: DE

Ref legal event code: R097

Ref document number: 602013036079

Country of ref document: DE

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: CZ

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180418

Ref country code: RO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180418

Ref country code: MC

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180418

Ref country code: EE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180418

Ref country code: SK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180418

PLBE No opposition filed within time limit

Free format text: ORIGINAL CODE: 0009261

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: NO OPPOSITION FILED WITHIN TIME LIMIT

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SM

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180418

26N No opposition filed

Effective date: 20190121

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LU

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20180531

REG Reference to a national code

Ref country code: NL

Ref legal event code: FP

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180418

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MT

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20180531

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: TR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180418

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: HU

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT; INVALID AB INITIO

Effective date: 20130531

Ref country code: MK

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20180418

Ref country code: CY

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180418

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180818

REG Reference to a national code

Ref country code: AT

Ref legal event code: UEP

Ref document number: 991041

Country of ref document: AT

Kind code of ref document: T

Effective date: 20180418

P01 Opt-out of the competence of the unified patent court (upc) registered

Effective date: 20230516

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: IT

Payment date: 20230523

Year of fee payment: 11

Ref country code: IE

Payment date: 20230522

Year of fee payment: 11

Ref country code: DK

Payment date: 20230524

Year of fee payment: 11

Ref country code: DE

Payment date: 20230519

Year of fee payment: 11

Ref country code: CH

Payment date: 20230605

Year of fee payment: 11

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: SE

Payment date: 20230519

Year of fee payment: 11

Ref country code: AT

Payment date: 20230522

Year of fee payment: 11

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: BE

Payment date: 20230519

Year of fee payment: 11

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: GB

Payment date: 20230524

Year of fee payment: 11

Ref country code: ES

Payment date: 20230725

Year of fee payment: 11

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: NL

Payment date: 20240415

Year of fee payment: 12

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: FR

Payment date: 20240328

Year of fee payment: 12

REG Reference to a national code

Ref country code: DE

Ref legal event code: R082

Ref document number: 602013036079

Country of ref document: DE

Ref country code: DE

Ref legal event code: R081

Ref document number: 602013036079

Country of ref document: DE

Owner name: NXERA PHARMA UK LIMITED, GREAT ABINGTON, GB

Free format text: FORMER OWNER: HEPTARES THERAPEUTICS LIMITED, WELWYN GARDEN CITY, HERTFORDSHIRE, GB